EP2620163A1 - Conjugués d'agoniste opioïde-oligomère - Google Patents

Conjugués d'agoniste opioïde-oligomère Download PDF

Info

Publication number
EP2620163A1
EP2620163A1 EP13155667.2A EP13155667A EP2620163A1 EP 2620163 A1 EP2620163 A1 EP 2620163A1 EP 13155667 A EP13155667 A EP 13155667A EP 2620163 A1 EP2620163 A1 EP 2620163A1
Authority
EP
European Patent Office
Prior art keywords
oligomer
compound
water
soluble
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP13155667.2A
Other languages
German (de)
English (en)
Other versions
EP2620163B1 (fr
Inventor
Jennifer Riggs-Sauthier
Bo-Liang Deng
Timothy Andrew Riley
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nektar Therapeutics
Original Assignee
Nektar Therapeutics
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nektar Therapeutics filed Critical Nektar Therapeutics
Priority to EP17151793.1A priority Critical patent/EP3222293B1/fr
Publication of EP2620163A1 publication Critical patent/EP2620163A1/fr
Application granted granted Critical
Publication of EP2620163B1 publication Critical patent/EP2620163B1/fr
Not-in-force legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4748Quinolines; Isoquinolines forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics

Definitions

  • This invention provides (among other things) chemically modified opioid agonists that possess certain advantages over opioid agonists lacking the chemical modification.
  • the chemically modified opioid agonists described herein relate to and/or have application(s) in (among others) the fields of drug discovery, pharmacotherapy, physiology, organic chemistry and polymer chemistry.
  • Opioid agonists such as morphine
  • Opioid agonists have long been used to treat patients suffering from pain.
  • Opioid agonists exert their analgesic and other pharmacological effects through interactions with opioid receptors, of which, there are three main classes: mu ( ⁇ ) receptors, kappa ( ⁇ ) receptors, and delta ( ⁇ ) receptors.
  • opioid receptors of which, there are three main classes: mu ( ⁇ ) receptors, kappa ( ⁇ ) receptors, and delta ( ⁇ ) receptors.
  • Most of the clinically used opioid agonists are relatively selective for mu receptors, although opioid agonists typically have agonist activity at other opioid receptors (particularly at increased concentrations).
  • Opioids exert their effects by selectively inhibiting the release of neurotransmitters, such as acetylcholine, norepinephrine, dopamine, serotonin, and substance P.
  • neurotransmitters such as acetylcholine, norepinephrine, dopamine, serotonin, and substance P.
  • opioid agonists represent an important class of agents employed in the management of pain.
  • the use of opioid agonists is associated with the potential for abuse.
  • oral administration of opioid agonists often results in significant first pass metabolism.
  • administration of opioid agonists results in significant CNS-mediated effects, such as slowed breathing, which can result in death.
  • CNS-mediated effects such as slowed breathing, which can result in death.
  • a reduction of any one of these or other characteristics would enhance their desirability as therapeutic drugs.
  • the present invention seeks to address these and other needs in the art by providing (among other things) a conjugate of a water-soluble, non-peptidic oligomer and a opioid agonist.
  • a compound comprising a residue of an opioid agonist covalently attached (preferably via a stable linkage) to a water-soluble, non-peptidic oligomer.
  • a compound comprising a residue of a kappa opioid agonist covalently attached (preferably via a stable linkage) to a water-soluble, non-peptidic oligomer [wherein it is understood that a kappa opioid agonist (i) is preferentially selective for kappa opioid receptors over both mu opioid receptors and delta opioid receptors within the same mammalian species, and (ii) will have agonist activity at the kappa receptor].
  • a compound comprising a residue of a mu opioid agonist covalently attached (preferably via a stable linkage) to a water-soluble, non-peptidic oligomer [wherein it is understood that a kappa opioid agonist (i) is preferentially selective for mu opioid receptors over both kappa opioid receptors and delta opioid receptors within the same mammalian species, and (ii) will have agonist activity at the mu receptor].
  • a compound comprising a residue of an opioid agonist covalently attached via a stable linkage to a water-soluble, non-peptidic oligomer, wherein the opioid agonist has a structure encompassed by the following formula: wherein:
  • a compound comprising a residue of an opioid agonist covalently attached via a stable or degradable linkage to a water-soluble, non-peptidic oligomer, wherein the opioid agonist is selected from the group consisting of asimadoline, bremazocine, enadoline, ethylketocyclazocine, GR89,696, ICI204448, ICI197067, PD117,302, nalbuphine, pentazocine, quadazocine (WIN 44,441-3), salvinorin A, spiradoline, TRK-820, U50488, and U69593.
  • the opioid agonist is selected from the group consisting of asimadoline, bremazocine, enadoline, ethylketocyclazocine, GR89,696, ICI204448, ICI197067, PD117,302, nalbuphine, pentazocine, quadazocine (WIN 44,441-3), salvinorin A, spiradoline
  • composition comprising:
  • a dosage form comprising a compound comprising a residue of an opioid agonist covalently attached via a stable linkage to a water-soluble, non-peptidic oligomer.
  • a method comprising covalently attaching a water-soluble, non-peptidic oligomer to an opioid agonist.
  • a method comprising administering a compound comprising a residue of an opioid agonist covalently attached via a stable linkage to a water-soluble, non-peptidic oligomer.
  • a method comprising binding (e.g., selectively binding) mu opioid receptors, wherein said binding is achieved by administering a compound comprising a residue of an opioid agonist covalently attached to a water-soluble, non-peptidic oligomer.
  • a method comprising binding (e.g., selectively binding) mu opioid receptors, wherein said binding is achieved by administering an effective amount to a mammalian patient a compound comprising a residue of an opioid agonist covalently attached to a water-soluble, non-peptidic oligomer.
  • a method comprising binding (e.g., selectively binding) kappa opioid receptors, wherein said binding is achieved by administering a compound comprising a residue of an opioid agonist covalently attached to a water-soluble, non-peptidic oligomer.
  • a method comprising binding (e.g., selectively binding) kappa opioid receptors, wherein said binding is achieved by administering an effective amount to a mammalian patient a compound comprising a residue of an opioid agonist covalently attached to a water-soluble, non-peptidic oligomer.
  • Water soluble, non-peptidic oligomer indicates an oligomer that is at least 35% (by weight) soluble, preferably greater than 70% (by weight), and more preferably greater than 95% (by weight) soluble, in water at room temperature.
  • an unfiltered aqueous preparation of a "water-soluble” oligomer transmits at least 75%, more preferably at least 95%, of the amount of light transmitted by the same solution after filtering. It is most preferred, however, that the water-soluble oligomer is at least 95% (by weight) soluble in water or completely soluble in water.
  • an oligomer is non-peptidic when it has less than 35% (by weight) of amino acid residues.
  • oligomer refers to one of the basic structural units of a polymer or oligomer.
  • a homo-oligomer a single repeating structural unit forms the oligomer.
  • two or more structural units are repeated -- either in a pattern or randomly -- to form the oligomer.
  • Preferred oligomers used in connection with present the invention are homo-oligomers.
  • the water-soluble, non-peptidic oligomer typically comprises one or more monomers serially attached to form a chain of monomers.
  • the oligomer can be formed from a single monomer type (i.e., is homo-oligomeric) or two or three monomer types (i.e., is co-oligomeric).
  • oligomer is a molecule possessing from about 2 to about 50 monomers, preferably from about 2 to about 30 monomers.
  • the architecture of an oligomer can vary.
  • Specific oligomers for use in the invention include those having a variety of geometries such as linear, branched, or forked, to be described in greater detail below.
  • PEG polyethylene glycol
  • polyethylene glycol is meant to encompass any water-soluble poly(ethylene oxide).
  • a "PEG oligomer” also called an oligoethylene glycol is one in which substantially all (and more preferably all) monomeric subunits are ethylene oxide subunits.
  • the oligomer may, however, contain distinct end capping moieties or functional groups, e.g., for conjugation.
  • PEG oligomers for use in the present invention will comprise one of the two following structures: "-(CH 2 CH 2 O) n -" or “-(CH 2 CH 2 O) n-1 CH 2 CH 2 -,” depending upon whether the terminal oxygen(s) has been displaced, e.g., during a synthetic transformation.
  • "n" varies from about 2 to 50, preferably from about 2 to about 30, and the terminal groups and architecture of the overall PEG can vary.
  • PEG further comprises a functional group, A, for linking to, e.g., a small molecule drug
  • the functional group when covalently attached to a PEG oligomer does not result in formation of (i) an oxygen-oxygen bond (-O-O-, a peroxide linkage), or (ii) a nitrogen-oxygen bond (N-O, ON).
  • An “end capping group” is generally a non-reactive carbon-containing group attached to a terminal oxygen of a PEG oligomer.
  • Exemplary end capping groups comprise a C 1-5 alkyl group, such as methyl, ethyl and benzyl), as well as aryl, heteroaryl, cyclo, heterocyclo, and the like.
  • the preferred capping groups have relatively low molecular weights such as methyl or ethyl.
  • the end-capping group can also comprise a detectable label.
  • Such labels include, without limitation, fluorescers, chemiluminescers, moieties used in enzyme labeling, colorimetric labels (e.g., dyes), metal ions, and radioactive moieties.
  • Branched in reference to the geometry or overall structure of an oligomer, refers to an oligomer having two or more polymers representing distinct “arms" that extend from a branch point.
  • Formked in reference to the geometry or overall structure of an oligomer, refers to an oligomer having two or more functional groups (typically through one or more atoms) extending from a branch point.
  • a "branch point” refers to a bifurcation point comprising one or more atoms at which an oligomer branches or forks from a linear structure into one or more additional arms.
  • reactive refers to a functional group that reacts readily or at a practical rate under conventional conditions of organic synthesis. This is in contrast to those groups that either do not react or require strong catalysts or impractical reaction conditions in order to react (i.e., a "nonreactive” or “inert” group).
  • a “protecting group” is a moiety that prevents or blocks reaction of a particular chemically reactive functional group in a molecule under certain reaction conditions.
  • the protecting group will vary depending upon the type of chemically reactive group being protected as well as the reaction conditions to be employed and the presence of additional reactive or protecting groups in the molecule.
  • Functional groups which may be protected include, by way of example, carboxylic acid groups, amino groups, hydroxyl groups, thiol groups, carbonyl groups and the like.
  • protecting groups for carboxylic acids include esters (such as a p-methoxybenzyl ester), amides and hydrazides; for amino groups, carbamates (such as tert-butoxycarbonyl) and amides; for hydroxyl groups, ethers and esters; for thiol groups, thioethers and thioesters; for carbonyl groups, acetals and ketals; and the like.
  • Such protecting groups are well-known to those skilled in the art and are described, for example, in T.W. Greene and G.M. Wuts, Protecting Groups in Organic Synthesis, Third Edition, Wiley, New York, 1999 , and references cited therein.
  • a functional group in "protected form” refers to a functional group bearing a protecting group.
  • the term “functional group” or any synonym thereof encompasses protected forms thereof.
  • a “physiologically cleavable” or “hydrolyzable” or “degradable” bond is a relatively labile bond that reacts with water (i.e., is hydrolyzed) under ordinary physiological conditions.
  • the tendency of a bond to hydrolyze in water under ordinary physiological conditions will depend not only on the general type of linkage connecting two central atoms but also on the substituents attached to these central atoms. Such bonds are generally recognizable by those of ordinary skill in the art.
  • Appropriate hydrolytically unstable or weak linkages include but are not limited to carboxylate ester, phosphate ester, anhydrides, acetals, ketals, acyloxyalkyl ether, imines, orthoesters, peptides, oligonucleotides, thioesters, and carbonates.
  • An “enzymatically degradable linkage” means a linkage that is subject to degradation by one or more enzymes under ordinary physiological conditions.
  • a “stable” linkage or bond refers to a chemical moiety or bond, typically a covalent bond, that is substantially stable in water, that is to say, does not undergo hydrolysis under ordinary physiological conditions to any appreciable extent over an extended period of time.
  • hydrolytically stable linkages include but are not limited to the following: carbon-carbon bonds (e.g., in aliphatic chains), ethers, amides, urethanes, amines, and the like.
  • a stable linkage is one that exhibits a rate of hydrolysis of less than about 1-2% per day under ordinary physiological conditions. Hydrolysis rates of representative chemical bonds can be found in most standard chemistry textbooks.
  • substantially or “essentially” means nearly totally or completely, for instance, 95% or greater, more preferably 97% or greater, still more preferably 98% or greater, even more preferably 99% or greater, yet still more preferably 99.9% or greater, with 99.99% or greater being most preferred of some given quantity.
  • “Monodisperse” refers to an oligomer composition wherein substantially all of the oligomers in the composition have a well-defined, single molecular weight and defined number of monomers, as determined by chromatography or mass spectrometry.
  • Monodisperse oligomer compositions are in one sense pure, that is, substantially comprising molecules having a single and definable number of monomers rather than several different numbers of monomers (i.e., an oligomer composition having three or more different oligomer sizes).
  • a monodisperse oligomer composition possesses a MW/Mn value of 1.0005 or less, and more preferably, a MW/Mn value of 1.0000.
  • a composition comprised of monodisperse conjugates means that substantially all oligomers of all conjugates in the composition have a single and definable number (as a whole number) of monomers rather than a distribution and would possess a MW/Mn value of 1.0005, and more preferably, a MW/Mn value of 1.0000 if the oligomer were not attached to the residue of the opioid agonist.
  • a composition comprised of monodisperse conjugates can include, however, one or more nonconjugate substances such as solvents, reagents, excipients, and so forth.
  • Bimodal in reference to an oligomer composition, refers to an oligomer composition wherein substantially all oligomers in the composition have one of two definable and different numbers (as whole numbers) of monomers rather than a distribution, and whose distribution of molecular weights, when plotted as a number fraction versus molecular weight, appears as two separate identifiable peaks.
  • each peak is generally symmetric about its mean, although the size of the two peaks may differ.
  • the polydispersity index of each peak in the bimodal distribution, Mw/Mn is 1.01 or less, more preferably 1.001 or less, and even more preferably 1.0005 or less, and most preferably a MW/Mn value of 1.0000.
  • a composition comprised of bimodal conjugates means that substantially all oligomers of all conjugates in the composition have one of two definable and different numbers (as whole numbers) of monomers rather than a large distribution and would possess a MW/Mn value of 1.01 or less, more preferably 1.001 or less and even more preferably 1.0005 or less, and most preferably a MW/Mn value of 1.0000 if the oligomer were not attached to the residue of the opioid agonist.
  • a composition comprised of bimodal conjugates can include, however, one or more nonconjugate substances such as solvents, reagents, excipients, and so forth.
  • opioid agonist is broadly used herein to refer to an organic, inorganic, or organometallic compound typically having a molecular weight of less than about 1000 Daltons (and typically less than 500 Daltons) and having some degree of activity as a mu and/or kappa agonist.
  • Opioid agonists encompass oligopeptides and other biomolecules having a molecular weight of less than about 1000.
  • a “biological membrane” is any membrane, typically made from specialized cells or tissues, that serves as a barrier to at least some foreign entities or otherwise undesirable materials.
  • a “biological membrane” includes those membranes that are associated with physiological protective barriers including, for example: the blood-brain barrier (BBB); the blood-cerebrospinal fluid barrier; the blood-placental barrier; the blood-milk barrier; the blood-testes barrier; and mucosal barriers including the vaginal mucosa, urethral mucosa, anal mucosa, buccal mucosa, sublingual mucosa, rectal mucosa, and so forth. Unless the context clearly dictates otherwise, the term “biological membrane” does not include those membranes associated with the middle gastro-intestinal tract (e.g., stomach and small intestines).
  • a "biological membrane crossing rate,” as used herein, provides a measure of a compound's ability to cross a biological membrane (such as the membrane associated with the blood-brain barrier).
  • a variety of methods can be used to assess transport of a molecule across any given biological membrane.
  • Methods to assess the biological membrane crossing rate associated with any given biological barrier are known in the art, described herein and/or in the relevant literature, and/or can be determined by one of ordinary skill in the art.
  • a “reduced rate of metabolism” in reference to the present invention refers to a measurable reduction in the rate of metabolism of a water-soluble oligomer-small molecule drug conjugate as compared to rate of metabolism of the small molecule drug not attached to the water-soluble oligomer (i.e., the small molecule drug itself) or a reference standard material.
  • the same “reduced rate of metabolism” is required except that the small molecule drug (or reference standard material) and the corresponding conjugate are administered orally.
  • Orally administered drugs are absorbed from the gastro-intestinal tract into the portal circulation and must pass through the liver prior to reaching the systemic circulation.
  • the degree of first pass metabolism can be measured by a number of different approaches. For instance, animal blood samples can be collected at timed intervals and the plasma or serum analyzed by liquid chromatography/mass spectrometry for metabolite levels. Other techniques for measuring a "reduced rate of metabolism" associated with the first pass metabolism and other metabolic processes are known in the art, described herein and/or in the relevant literature, and/or can be determined by one of ordinary skill in the art.
  • a conjugate of the invention can provide a reduced rate of metabolism reduction satisfying at least one of the following values: at least about 30%; at least about 40%; at least about 50%; at least about 60%; at least about 70%; at least about 80%; and at least about 90%.
  • a compound (such as a small molecule drug or conjugate thereof) that is "orally bioavailable" is one that preferably possesses a bioavailability when administered orally of greater than 25%, and preferably greater than 70%, where a compound's bioavailability is the fraction of administered drug that reaches the systemic circulation in unmetabolized form.
  • Alkyl refers to a hydrocarbon chain, typically ranging from about 1 to 20 atoms in length. Such hydrocarbon chains are preferably but not necessarily saturated and may be branched or straight chain, although typically straight chain is preferred. Exemplary alkyl groups include methyl, ethyl, propyl, butyl, pentyl, 1-methylbutyl, 1-ethylpropyl, 3-methylpentyl, and the like. As used herein, “alkyl” includes cycloalkyl when three or more carbon atoms are referenced. An “alkenyl” group is an alkyl of 2 to 20 carbon atoms with at least one carbon-carbon double bond.
  • substituted alkyl or “substituted C q-r alkyl” where q and r are integers identifying the range of carbon atoms contained in the alkyl group, denotes the above alkyl groups that are substituted by one, two or three halo (e.g., F, Cl, Br, I), trifluoromethyl, hydroxy, C 1-7 alkyl (e.g., methyl, ethyl, n-propyl, isopropyl, butyl, t-butyl, and so forth), C 1-7 alkoxy, C 1-7 acyloxy, C 3-7 heterocyclic, amino, phenoxy, nitro, carboxy, carboxy, acyl, cyano.
  • the substituted alkyl groups may be substituted once, twice or three times with the same or with different substituents.
  • “Lower alkyl” refers to an alkyl group containing from 1 to 6 carbon atoms, and may be straight chain or branched, as exemplified by methyl, ethyl, n-butyl, i-butyl, t-butyl.
  • “Lower alkenyl” refers to a lower alkyl group of 2 to 6 carbon atoms having at least one carbon-carbon double bond.
  • Non-interfering substituents are those groups that, when present in a molecule, are typically non-reactive with other functional groups contained within the molecule.
  • Alkoxy refers to an -O-R group, wherein R is alkyl or substituted alkyl, preferably C 1 -C 20 alkyl (e.g., methoxy, ethoxy, propyloxy, benzyl, etc.), preferably C 1- C 7 .
  • “Pharmaceutically acceptable excipient” or “pharmaceutically acceptable carrier” refers to component that can be included in the compositions of the invention in order to provide for a composition that has an advantage (e.g., more suited for administration to a patient) over a composition lacking the component and that is recognized as not causing significant adverse toxicological effects to a patient.
  • aryl means an aromatic group having up to 14 carbon atoms.
  • Aryl groups include phenyl, naphthyl, biphenyl, phenanthrenyl, naphthacenyl, and the like.
  • Substituted phenyl and “substituted aryl” denote a phenyl group and aryl group, respectively, substituted with one, two, three, four or five (e.g.
  • halo F, Cl, Br, I
  • hydroxy hydroxy
  • cyano nitro
  • alkyl e.g., C 1-6 alkyl
  • alkoxy e.g., C 1-6 alkoxy
  • benzyloxy carboxy, aryl, and so forth.
  • aromatic-containing moiety is a collection of atoms containing at least aryl and optionally one or more atoms. Suitable aromatic-containing moieties are described herein.
  • an "alkyl” moiety generally refers to a monovalent radical (e.g., CH 3 -CH 2 -)
  • a bivalent linking moiety can be "alkyl,” in which case those skilled in the art will understand the alkyl to be a divalent radical (e.g., -CH 2 -CH 2 -), which is equivalent to the term “alkylene.”
  • alkylene e.g., -CH 2 -CH 2 -
  • aryl refers to the corresponding divalent moiety, arylene. All atoms are understood to have their normal number of valences for bond formation (i.e., 4 for carbon, 3 for N, 2 for O, and 2, 4, or 6 for S, depending on the oxidation state of the S).
  • “Pharmacologically effective amount,” “physiologically effective amount,” and “therapeutically effective amount” are used interchangeably herein to mean the amount of a water-soluble oligomer-small molecule drug conjugate present in a composition that is needed to provide a threshold level of active agent and/or conjugate in the bloodstream or in the target tissue.
  • the precise amount will depend upon numerous factors, e.g., the particular active agent, the components and physical characteristics of the composition, intended patient population, patient considerations, and the like, and can readily be determined by one skilled in the art, based upon the information provided herein and available in the relevant literature.
  • a “difunctional" oligomer is an oligomer having two functional groups contained therein, typically at its termini. When the functional groups are the same, the oligomer is said to be homodifunctional. When the functional groups are different, the oligomer is said to be heterobifunctional.
  • a basic reactant or an acidic reactant described herein include neutral, charged, and any corresponding salt forms thereof.
  • patient refers to a living organism suffering from or prone to a condition that can be prevented or treated by administration of a conjugate as described herein, typically, but not necessarily, in the form of a water-soluble oligomer-small molecule drug conjugate, and includes both humans and animals.
  • the present invention is directed to (among other things) a compound comprising a residue of an opioid agonist covalently attached via a stable or degradable linkage to a water-soluble, non-peptidic oligomer.
  • a compound comprising a residue of an opioid agonist covalently attached via a stable or degradable linkage to a water-soluble, non-peptidic oligomer, wherein the opioid agonist has a structure encompassed by the following formula: wherein:
  • Examples of specific opioid agonists include those selected from the group consisting acetorphine, acetyldihydrocodeine, acetyldihydrocodeinone, acetylmorphinone, alfentanil, allylprodine, alphaprodine, anileridine, benzylmorphine, bezitramide, buprenorphine, butorphanol, clonitazene, codeine, desomorphine, dextromoramide, dezocine, diampromide, diamorphone, dihydrocodeine, dihydromorphine, dimenoxadol, dimepheptanol, dimethylthiambutene, dioxaphetyl butyrate, dipipanone, eptazocine, ethoheptazine, ethylmethylthiambutene, ethylmorphine, etonitazene, etorphine, dihydroet
  • the opioid agonist is selected from the group consisting of hydrocodone, morphine, hydromorphone, oxycodone, codeine, levorphanol, meperidine, methadone, oxymorphone, buprenorphine, fentanyl, dipipanone, heroin, tramadol, nalbuphine, etorphine, dihydroetorphine, butorphanol, levorphanol.
  • an advantage of the compounds of the present invention is their ability to retain some degree of opioid agonist activity while also exhibiting a decrease in metabolism and/or resulting in a decrease of CNS-mediated effects associated with the corresponding opioid agonist in unconjugated form.
  • the oligomer-containing conjugates described herein -- in contrast to the unconjugated "original" opioid agonist -- are not metabolized as readily because the oligomer serves to reduce the overall affinity of the compound to substrates that can metabolize opioid agonists.
  • the extra size introduced by the oligomer -- in contrast to the unconjugated "original” opioid agonist -- reduces the ability of the compound to cross the blood-brain barrier.
  • oligomers e.g., from a monodisperse or bimodal composition of oligomers, in contrast to relatively impure compositions
  • a conjugate of the invention when administered by any of a number of suitable administration routes, such as parenteral, oral, transdermal, buccal, pulmonary, or nasal, exhibits reduced penetration across the blood-brain barrier. It is preferred that the conjugate exhibit slowed, minimal or effectively no crossing of the blood-brain barrier, while still crossing the gastro-intestinal (GI) walls and into the systemic circulation if oral delivery is intended.
  • the conjugates of the invention maintain a degree of bioactivity as well as bioavailability in their conjugated form in comparison to the bioactivity and bioavailability of the compound free of all oligomers.
  • this barrier restricts the transport of drugs from the blood to the brain.
  • This barrier consists of a continuous layer of unique endothelial cells joined by tight junctions.
  • the cerebral capillaries which comprise more than 95% of the total surface area of the BBB, represent the principal route for the entry of most solutes and drugs into the central nervous system.
  • RBP in situ rat brain perfusion
  • a physiologic buffer containing the analyte (typically but not necessarily at a 5 micromolar concentration level) is perfused at a flow rate of about 10 mL/minute in a single pass perfusion experiment. After 30 seconds, the perfusion is stopped and the brain vascular contents are washed out with compound-free buffer for an additional 30 seconds. The brain tissue is then removed and analyzed for compound concentrations via liquid chromatograph with tandem mass spectrometry detection (LC/MS/MS). Alternatively, blood-brain barrier permeability can be estimated based upon a calculation of the compound's molecular polar surface area ("PSA”), which is defined as the sum of surface contributions of polar atoms (usually oxygens, nitrogens and attached hydrogens) in a molecule.
  • PSA molecular polar surface area
  • the PSA has been shown to correlate with compound transport properties such as blood-brain barrier transport.
  • Methods for determining a compound's PSA can be found, e.g., in, Ertl, P., et al., J. Med. Chem. 2000, 43, 3714-3717 ; and Kelder, J., et al., Pharm. Res. 1999, 16, 1514-1519 .
  • the water-soluble, non-peptidic oligomer-small molecule drug conjugate exhibits a blood-brain barrier crossing rate that is reduced as compared to the crossing rate of the small molecule drug not attached to the water-soluble, non-peptidic oligomer.
  • Preferred exemplary reductions in blood-brain barrier crossing rates for the compounds described herein include reductions of: at least about 30%; at least about 40%; at least about 50%; at least about 60%; at least about 70%; at least about 80%; or at least about 90%, when compared to the blood-brain barrier crossing rate of the small molecule drug not attached to the water-soluble oligomer.
  • a preferred reduction in the blood-brain barrier crossing rate for a conjugate is at least about 20%.
  • the compounds of the invention include a residue of an opioid agonist.
  • Assays for determining whether a given compound (regardless of whether the compound is in conjugated form or not) can act as an agonist on a mu receptor or a kappa receptors are described infra.
  • opioid agonists can be obtained from commercial sources.
  • opioid agonists can be obtained through chemical synthesis. Synthetic approaches for preparing opioid agonists are described in the literature and in, for example, U.S. Patent Nos.: 2,628,962 , 2,654,756 , 2,649,454 , and 2,806,033 .
  • Each of these (and other) opioid agonists can be covalently attached (either directly or through one or more atoms) to a water-soluble, non-peptidic oligomer.
  • Small molecule drugs useful in the invention generally have a molecular weight of less than 1000 Da.
  • Exemplary molecular weights of small molecule drugs include molecular weights of: less than about 950; less than about 900; less than about 850; less than about 800; less than about 750; less than about 700; less than about 650; less than about 600; less than about 550; less than about 500; less than about 450; less than about 400; less than about 350; and less than about 300.
  • the small molecule drug used in the invention may be in a racemic mixture, or an optically active form, for example, a single optically active enantiomer, or any combination or ratio of enantiomers (i.e., scalemic mixture).
  • the small molecule drug may possess one or more geometric isomers.
  • geometric isomers a composition can comprise a single geometric isomer or a mixture of two or more geometric isomers.
  • a small molecule drug for use in the present invention can be in its customary active form, or may possess some degree of modification.
  • a small molecule drug may have a targeting agent, tag, or transporter attached thereto, prior to or after covalent attachment of an oligomer.
  • the small molecule drug may possess a lipophilic moiety attached thereto, such as a phospholipid (e.g., distearoylphosphatidylethanolamine or "DSPE,” dipalmitoylphosphatidylethanolamine or "DPPE,” and so forth) or a small fatty acid.
  • a phospholipid e.g., distearoylphosphatidylethanolamine or "DSPE,” dipalmitoylphosphatidylethanolamine or "DPPE,” and so forth
  • DPPE dipalmitoylphosphatidylethanolamine
  • the small molecule drug moiety does not include attachment to a lipophilic moiety.
  • the opioid agonist for coupling to a water-soluble, non-peptidic oligomer possesses a free hydroxyl, carboxyl, thio, amino group, or the like (i.e., "handle") suitable for covalent attachment to the oligomer.
  • the opioid agonist can be modified by introduction of a reactive group, preferably by conversion of one of its existing functional groups to a functional group suitable for formation of a stable covalent linkage between the oligomer and the drug.
  • each oligomer is composed of up to three different monomer types selected from the group consisting of: alkylene oxide, such as ethylene oxide or propylene oxide; olefinic alcohol, such as vinyl alcohol, 1-propenol or 2-propenol; vinyl pyrrolidone; hydroxyalkyl methacrylamide or hydroxyalkyl methacrylate, where alkyl is preferably methyl; ⁇ -hydroxy acid, such as lactic acid or glycolic acid; phosphazene, oxazoline, amino acids, carbohydrates such as monosaccharides, saccharide or mannitol; and N-acryloylmorpholine.
  • alkylene oxide such as ethylene oxide or propylene oxide
  • olefinic alcohol such as vinyl alcohol, 1-propenol or 2-propenol
  • vinyl pyrrolidone hydroxyalkyl methacrylamide or hydroxyalkyl methacrylate, where alkyl is preferably methyl
  • ⁇ -hydroxy acid such as lactic acid or
  • Preferred monomer types include alkylene oxide, olefinic alcohol, hydroxyalkyl methacrylamide or methacrylate, N-acryloylmorpholine, and ⁇ -hydroxy acid.
  • each oligomer is, independently, a co-oligomer of two monomer types selected from this group, or, more preferably, is a homo-oligomer of one monomer type selected from this group.
  • the two monomer types in a co-oligomer may be of the same monomer type, for example, two alkylene oxides, such as ethylene oxide and propylene oxide.
  • the oligomer is a homo-oligomer of ethylene oxide.
  • the terminus (or termini) of the oligomer that is not covalently attached to a small molecule is capped to render it unreactive.
  • the terminus may include a reactive group. When the terminus is a reactive group, the reactive group is either selected such that it is unreactive under the conditions of formation of the final oligomer or during covalent attachment of the oligomer to a small molecule drug, or it is protected as necessary.
  • One common end-functional group is hydroxyl or -OH, particularly for oligoethylene oxides.
  • the water-soluble, non-peptidic oligomer (e.g., "POLY" in various structures provided herein) can have any of a number of different geometries. For example, it can be linear, branched, or forked. Most typically, the water-soluble, non-peptidic oligomer is linear or is branched, for example, having one branch point. Although much of the discussion herein is focused upon poly(ethylene oxide) as an illustrative oligomer, the discussion and structures presented herein can be readily extended to encompass any of the water-soluble, non-peptidic oligomers described above.
  • the molecular weight of the water-soluble, non-peptidic oligomer, excluding the linker portion, is generally relatively low.
  • Exemplary values of the molecular weight of the water-soluble polymer include: below about 1500; below about 1450; below about 1400; below about 1350; below about 1300; below about 1250; below about 1200; below about 1150; below about 1100; below about 1050; below about 1000; below about 950; below about 900; below about 850; below about 800; below about 750; below about 700; below about 650; below about 600; below about 550; below about 500; below about 450; below about 400; below about 350; below about 300; below about 250; below about 200; and below about 100 Daltons.
  • Exemplary ranges of molecular weights of the water-soluble, non-peptidic oligomer include: from about 100 to about 1400 Daltons; from about 100 to about 1200 Daltons; from about 100 to about 800 Daltons; from about 100 to about 500 Daltons; from about 100 to about 400 Daltons; from about 200 to about 500 Daltons; from about 200 to about 400 Daltons; from about 75 to 1000 Daltons; and from about 75 to about 750 Daltons.
  • the number of monomers in the water-soluble, non-peptidic oligomer falls within one or more of the following ranges: between about 1 and about 30 (inclusive); between about 1 and about 25; between about 1 and about 20; between about 1 and about 15; between about 1 and about 12; between about 1 and about 10.
  • the number of monomers in series in the oligomer (and the corresponding conjugate) is one of 1, 2, 3, 4, 5, 6, 7, or 8.
  • the oligomer (and the corresponding conjugate) contains 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 monomers.
  • the oligomer (and the corresponding conjugate) possesses 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30 monomers in series.
  • n is an integer that can be 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29 or 30, and can fall within one or more of the following ranges: between about 1 and about 25; between about 1 and about 20; between about 1 and about 15; between about 1 and about 12; between about 1 and about 10.
  • the water-soluble, non-peptidic oligomer has 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 monomers, these values correspond to a methoxy end-capped oligo(ethylene oxide) having a molecular weights of about 75, 119, 163, 207, 251, 295, 339, 383, 427, and 471 Daltons, respectively.
  • the oligomer has 11, 12, 13, 14, or 15 monomers, these values correspond to methoxy end-capped oligo(ethylene oxide) having molecular weights corresponding to about 515, 559, 603, 647, and 691 Daltons, respectively.
  • the composition containing an activated form of the water-soluble, non-peptidic oligomer be monodispersed.
  • the composition will possess a bimodal distribution centering around any two of the above numbers of monomers.
  • the polydispersity index of each peak in the bimodal distribution, Mw/Mn is 1.01 or less, and even more preferably, is 1.001 or less, and even more preferably is 1.0005 or less.
  • each peak possesses a MW/Mn value of 1.0000.
  • a bimodal oligomer may have any one of the following exemplary combinations of monomer subunits: 1-2, 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 1-9, 1-10, and so forth; 2-3, 2-4, 2-5, 2-6, 2-7, 2-8, 2-9, 2-10, and so forth; 3-4, 3-5, 3-6, 3-7, 3-8, 3-9, 3-10, and so forth; 4-5, 4-6, 4-7, 4-8, 4-9, 4-10, and so forth; 5-6, 5-7, 5-8, 5-9, 5-10, and so forth; 6-7, 6-8, 6-9, 6-10, and so forth; 7-8, 7-9, 7-10, and so forth; and 8-9, 8-10, and so forth.
  • the composition containing an activated form of the water-soluble, non-peptidic oligomer will be trimodal or even tetramodal, possessing a range of monomers units as previously described.
  • Oligomer compositions possessing a well-defined mixture of oligomers i.e., being bimodal, trimodal, tetramodal, and so forth
  • can be prepared by mixing purified monodisperse oligomers to obtain a desired profile of oligomers a mixture of two oligomers differing only in the number of monomers is bimodal; a mixture of three oligomers differing only in the number of monomers is trimodal; a mixture of four oligomers differing only in the number of monomers is tetramodal
  • a desired profile of oligomers a mixture of two oligomers differing only in the number of monomers is bimodal; a mixture of three oligomers differing only in the number of monomers is trimodal; a mixture of four oligomers differing only in the
  • the water-soluble, non-peptidic oligomer is obtained from a composition that is preferably unimolecular or monodisperse. That is, the oligomers in the composition possess the same discrete molecular weight value rather than a distribution of molecular weights.
  • Some monodisperse oligomers can be purchased from commercial sources such as those available from Sigma-Aldrich, or alternatively, can be prepared directly from commercially available starting materials such as Sigma-Aldrich.
  • Water-soluble, non-peptidic oligomers can be prepared as described in Chen Y., Baker, G.L., J. Org. Chem., 6870-6873 (1999 ), WO 02/098949 , and U.S. Patent Application Publication 2005/0136031 .
  • the spacer moiety (through which the water-soluble, non-peptidic polymer is attached to the opioid agonist) may be a single bond, a single atom, such as an oxygen atom or a sulfur atom, two atoms, or a number of atoms.
  • a spacer moiety is typically but is not necessarily linear in nature.
  • the spacer moiety, "X" is preferably hydrolytically stable, and is preferably also enzymatically stable.
  • the spacer moiety "X" is one having a chain length of less than about 12 atoms, and preferably less than about 10 atoms, and even more preferably less than about 8 atoms and even more preferably less than about 5 atoms, whereby length is meant the number of atoms in a single chain, not counting substituents.
  • the spacer moiety linkage does not comprise further spacer groups.
  • the spacer moiety "X" comprises an ether, amide, urethane, amine, thioether, urea, or a carbon-carbon bond. Functional groups such as those discussed below, and illustrated in the examples, are typically used for forming the linkages.
  • the spacer moiety may less preferably also comprise (or be adjacent to or flanked by) spacer groups, as described further below.
  • a spacer moiety, X may be any of the following: "-" (i.e., a covalent bond, that may be stable or degradable, between the residue of the small molecule opioid agonist and the water-soluble, non-peptidic oligomer), -O-, -NH-, -S-, -C(O)-, C(O)-NH, NH-C(O)-NH, O-C(O)-NH, -C(S)-, -CH 2 -, -CH 2 -CH 2 -, -CH 2 -CH 2 -CH 2 -, -CH 2 -CH 2 -CH 2 -, -CH 2 -CH 2 -CH 2 -CH 2 -, -O-CH 2 -, -CH 2 -O-, -O-CH 2 -CH 2 -, -CH 2 -O-CH 2 -, -CH 2 -O-CH 2 -, -CH 2 -O
  • a group of atoms is not considered a spacer moiety when it is immediately adjacent to an oligomer segment, and the group of atoms is the same as a monomer of the oligomer such that the group would represent a mere extension of the oligomer chain.
  • the linkage "X" between the water-soluble, non-peptidic oligomer and the small molecule is typically formed by reaction of a functional group on a terminus of the oligomer (or one or more monomers when it is desired to "grow” the oligomer onto the opioid agonist) with a corresponding functional group within the opioid agonist.
  • Illustrative reactions are described briefly below. For example, an amino group on an oligomer may be reacted with a carboxylic acid or an activated carboxylic acid derivative on the small molecule, or vice versa, to produce an amide linkage.
  • reaction of an amine on an oligomer with an activated carbonate e.g.
  • succinimidyl or benzotriazyl carbonate on the drug, or vice versa, forms a carbamate linkage.
  • reaction of an alcohol (alkoxide) group on an oligomer with an alkyl halide, or halide group within a drug, or vice versa forms an ether linkage.
  • a small molecule having an aldehyde function is coupled to an oligomer amino group by reductive amination, resulting in formation of a secondary amine linkage between the oligomer and the small molecule.
  • a particularly preferred water-soluble, non-peptidic oligomer is an oligomer bearing an aldehyde functional group.
  • the oligomer will have the following structure: CH 3 O-(CH 2 -CH 2 -O) n -(CH 2 ) p -C(O)H, wherein (n) is one of 1, 2, 3, 4, 5, 6, 7, 8, 9 and 10 and (p) is one of 1, 2, 3, 4, 5, 6 and 7.
  • Preferred (n) values include 3, 5 and 7 and preferred (p) values 2, 3 and 4.
  • the carbon atom alpha to the -C(O)H moiety can optionally be substituted with alkyl.
  • the terminus of the water-soluble, non-peptidic oligomer not bearing a functional group is capped to render it unreactive.
  • that group is either selected such that it is unreactive under the conditions of formation of the linkage "X,” or it is protected during the formation of the linkage "X.”
  • the water-soluble, non-peptidic oligomer includes at least one functional group prior to conjugation.
  • the functional group typically comprises an electrophilic or nucleophilic group for covalent attachment to a small molecule, depending upon the reactive group contained within or introduced into the small molecule.
  • nucleophilic groups that may be present in either the oligomer or the small molecule include hydroxyl, amine, hydrazine (-NHNH 2 ), hydrazide (-C(O)NHNH 2 ), and thiol.
  • Preferred nucleophiles include amine, hydrazine, hydrazide, and thiol, particularly amine.
  • Most small molecule drugs for covalent attachment to an oligomer will possess a free hydroxyl, amino, thio, aldehyde, ketone, or carboxyl group.
  • electrophilic functional groups that may be present in either the oligomer or the small molecule include carboxylic acid, carboxylic ester, particularly imide esters, orthoester, carbonate, isocyanate, isothiocyanate, aldehyde, ketone, thione, alkenyl, acrylate, methacrylate, acrylamide, sulfone, maleimide, disulfide, iodo, epoxy, sulfonate, thiosulfonate, silane, alkoxysilane, and halosilane.
  • succinimidyl ester or carbonate imidazoyl ester or carbonate, benzotriazole ester or carbonate
  • vinyl sulfone chloroethylsulfone
  • vinylpyridine pyridyl disulfide
  • iodoacetamide glyoxal
  • dione mesylate, tosylate, and tresylate (2,2,2-trifluoroethanesulfonate.
  • sulfur analogs of several of these groups such as thione, thione hydrate, thioketal, is 2-thiazolidine thione, etc., as well as hydrates or protected derivatives of any of the above moieties (e.g. aldehyde hydrate, hemiacetal, acetal, ketone hydrate, hemiketal, ketal, thioketal, thioacetal).
  • an "activated derivative" of a carboxylic acid refers to a carboxylic acid derivative which reacts readily with nucleophiles, generally much more readily than the underivatized carboxylic acid.
  • Activated carboxylic acids include, for example, acid halides (such as acid chlorides), anhydrides, carbonates, and esters.
  • esters include imide esters, of the general form -(CO)O-N[(CO)-] 2 ; for example, N-hydroxysuccinimidyl (NHS) esters or N-hydroxyphthalimidyl esters.
  • imidazolyl esters and benzotriazole esters Particularly preferred are activated propionic acid or butanoic acid esters, as described in co-owned U.S.
  • electrophilic groups include succinimidyl carbonate, maleimide, benzotriazole carbonate, glycidyl ether, imidazoyl carbonate, p-nitrophenyl carbonate, acrylate, tresylate, aldehyde, and orthopyridyl disulfide.
  • electrophilic groups are subject to reaction with nucleophiles, e.g. hydroxy, thio, or amino groups, to produce various bond types.
  • Preferred for the present invention are reactions which favor formation of a hydrolytically stable linkage.
  • carboxylic acids and activated derivatives thereof which include orthoesters, succinimidyl esters, imidazolyl esters, and benzotriazole esters, react with the above types of nucleophiles to form esters, thioesters, and amides, respectively, of which amides are the most hydrolytically stable.
  • Isocyanates react with hydroxyl or amino groups to form, respectively, carbamate (RNH-C(O)-OR') or urea (RNH-C(O)-NHR') linkages.
  • Aldehydes, ketones, glyoxals, diones and their hydrates or alcohol adducts i.e. aldehyde hydrate, hemiacetal, acetal, ketone hydrate, hemiketal, and ketal
  • aldehyde hydrate, hemiacetal, acetal, ketone hydrate, hemiketal, and ketal are preferably reacted with amines, followed by reduction of the resulting imine, if desired, to provide an amine linkage (reductive amination).
  • electrophilic functional groups include electrophilic double bonds to which nucleophilic groups, such as thiols, can be added, to form, for example, thioether bonds.
  • nucleophilic groups such as thiols
  • These groups include maleimides, vinyl sulfones, vinyl pyridine, acrylates, methacrylates, and acrylamides.
  • Other groups comprise leaving groups that can be displaced by a nucleophile; these include chloroethyl sulfone, pyridyl disulfides (which include a cleavable S-S bond), iodoacetamide, mesylate, tosylate, thiosulfonate, and tresylate.
  • Epoxides react by ring opening by a nucleophile, to form, for example, an ether or amine bond. Reactions involving complementary reactive groups such as those noted above on the oligomer and the small molecule are utilized to prepare the conjugates of the invention.
  • the opioid agonist may not have a functional group suited for conjugation.
  • the opioid agonist has an amide group, but an amine group is desired, it is possible to modify the amide group to an amine group by way of a Hofmann rearrangement, Curtius rearrangement (once the amide is converted to an azide) or Lossen rearrangement (once amide is concerted to hydroxamide followed by treatment with tolyene-2-sulfonyl chloride/base).
  • a conjugate of small molecule opioid agonist bearing a carboxyl group wherein the carboxyl group-bearing small molecule opioid agonist is coupled to an amino-terminated oligomeric ethylene glycol to provide a conjugate having an amide group covalently linking the small molecule opioid agonist to the oligomer.
  • This can be performed, for example, by combining the carboxyl group-bearing small molecule opioid agonist with the amino-terminated oligomeric ethylene glycol in the presence of a coupling reagent, (such as dicyclohexylcarbodiimide or "DCC”) in an anhydrous organic solvent.
  • a coupling reagent such as dicyclohexylcarbodiimide or "DCC”
  • a conjugate of a small molecule opioid agonist bearing a hydroxyl group wherein the hydroxyl group-bearing small molecule opioid agonist is coupled to an oligomeric ethylene glycol halide to result in an ether (-O-) linked small molecule conjugate.
  • This can be performed, for example, by using sodium hydride to deprotonate the hydroxyl group followed by reaction with a halide-terminated oligomeric ethylene glycol.
  • a conjugate of a small molecule opioid agonist bearing an amine group it is possible to prepare a conjugate of a small molecule opioid agonist bearing an amine group.
  • the amine group-bearing small molecule opioid agonist and an aldehyde-bearing oligomer are dissolved in a suitable buffer after which a suitable reducing agent (e.g., NaCNBH 3 ) is added.
  • a suitable reducing agent e.g., NaCNBH 3
  • a carboxylic acid-bearing oligomer and the amine group-bearing small molecule opioid agonist are combined, typically in the presence of a coupling reagent (e.g., DCC).
  • a coupling reagent e.g., DCC
  • Exemplary conjugates of the opioid agonists of Formula I include those having the following structure: wherein each of R 2 , R 3 , R 4 , the dotted line ("---"), Y 1 and R 5 is as previously defined with respect to Formula I, X is a spacer moiety and POLY is a water-soluble, non-peptidic oligomer.
  • Additional exemplary conjugates of the opioid agonists of Formula I include those having the following structure: wherein each of R 1 , R 2 , R 3 , R 4 , the dotted line ("---"), and Y 1 is as previously defined with respect to Formula I, X is a spacer moiety and POLY is a water-soluble, non-peptidic oligomer.
  • exemplary conjugates of the opioid agonists of Formula I include those having the following structure: wherein each of R 1 , R 2 , R 3 , R 4 , Y 1 and R 5 is as previously defined with respect to Formula I, X is a spacer moiety and POLY is a water-soluble, non-peptidic oligomer.
  • Still further exemplary conjugates of the opioid agonists of Formula I include those having the following structure: wherein each of R 1 , R 2 , R 3 , R 4 , Y 1 and R 5 is as previously defined with respect to Formula I, X is a spacer moiety and POLY is a water-soluble, non-peptidic oligomer.
  • Additional exemplary conjugates of the opioid agonists of Formula I include those having the following structure: wherein each of R 1 , R 3 , R 4 , the dotted line ("---"), Y 1 and R 5 is as previously defined with respect to Formula I, X is a spacer moiety and POLY is a water-soluble, non-peptidic oligomer.
  • Additional conjugates include those provided below: wherein, for each of the above conjugates, X is a linker (e.g., a covalent bond "-" or one or more atoms) and POLY is a water-soluble, non-peptidic oligomer.
  • X is a linker (e.g., a covalent bond "-" or one or more atoms) and POLY is a water-soluble, non-peptidic oligomer.
  • the conjugates of the invention can exhibit a reduced blood-brain barrier crossing rate. Moreover, the conjugates maintain at least about 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, or more of the bioactivity of the unmodified parent small molecule drug.
  • an optimally sized oligomer can be determined as follows.
  • an oligomer obtained from a monodisperse or bimodal water soluble oligomer is conjugated to the small molecule drug.
  • the drug is orally bioavailable, and on its own, exhibits a non-negligible blood-brain barrier crossing rate.
  • the ability of the conjugate to cross the blood-brain barrier is determined using an appropriate model and compared to that of the unmodified parent drug. If the results are favorable, that is to say, if, for example, the rate of crossing is significantly reduced, then the bioactivity of conjugate is further evaluated.
  • the compounds according to the invention maintain a significant degree of bioactivity relative to the parent drug, i.e., greater than about 30% of the bioactivity of the parent drug, or even more preferably, greater than about 50% of the bioactivity of the parent drug.
  • oligomer size By making small, incremental changes in oligomer size, and utilizing an experimental design approach, one can effectively identify a conjugate having a favorable balance of reduction in biological membrane crossing rate, bioactivity, and oral bioavailability. In some instances, attachment of an oligomer as described herein is effective to actually increase oral bioavailability of the drug.
  • one of ordinary skill in the art using routine experimentation, can determine a best suited molecular size and linkage for improving oral bioavailability by first preparing a series of oligomers with different weights and functional groups and then obtaining the necessary clearance profiles by administering the conjugates to a patient and taking periodic blood and/or urine sampling. Once a series of clearance profiles have been obtained for each tested conjugate, a suitable conjugate can be identified.
  • Animal models can also be used to study oral drug transport.
  • non- in vivo methods include rodent everted gut excised tissue and Caco-2 cell monolayer tissue-culture models. These models are useful in predicting oral drug bioavailability.
  • K D binding affinity
  • B max receptor number
  • human mu receptors can be recombinantly expressed in Chinese hamster ovary cells.
  • the radioligand [ 3 H]-diprenorphine (30-50 Ci/mmol) with a final ligand concentration of [0.3 nM] can be used.
  • Naloxone is used as a non-specific determinate [3.0 nM], a reference compound and positive control. Reactions are carried out in 50 mM TRIS-HCl (pH 7.4) containing 5 mM MgCl 2 , at 25 °C for 150 minutes. The reaction is terminated by rapid vacuum filtration onto glass fiber filters. Radioactivity trapped onto filters is determined and compared to control values in order to ascertain any interactions of test compound with the cloned mu binding site.
  • kappa opioid receptor agonist Similar testing can be performed for kappa opioid receptor agonist. See, for example, Lahti et al. (1985) Eur. Jrnl. Pharmac. 109:281-284 ; Rothman et al. (1992) Peptides 13:977-987 ; Kinouchi et al. (1991) Eur. Jrnl. Pharmac. 207:135-141 . Briefly, human kappa receptors can be obtained from guinea pig cerebellar membranes. The radioligand [ 3 H]-U-69593 (40-60 Ci/mmol) with a final ligand concentration of [0.75 nM] can be used.
  • U-69593 is used as a non-specific determinate [1.0 ⁇ M], a reference compound and positive control. Reactions are carried out in 50 mM HEPES (pH 7.4) at 30 °C for 120 minutes. The reaction is terminated by rapid vacuum filtration onto glass fiber filters. Radioactivity trapped onto filters is determined and compared to control values in order to ascertain any interactions of test compound with the cloned kappa binding site.
  • the present invention also includes pharmaceutical preparations comprising a conjugate as provided herein in combination with a pharmaceutical excipient.
  • the conjugate itself will be in a solid form (e.g., a precipitate), which can be combined with a suitable pharmaceutical excipient that can be in either solid or liquid form.
  • Exemplary excipients include, without limitation, those selected from the group consisting of carbohydrates, inorganic salts, antimicrobial agents, antioxidants, surfactants, buffers, acids, bases, and combinations thereof.
  • a carbohydrate such as a sugar, a derivatized sugar such as an alditol, aldonic acid, an esterified sugar, and/or a sugar polymer may be present as an excipient.
  • Specific carbohydrate excipients include, for example: monosaccharides, such as fructose, maltose, galactose, glucose, D-mannose, sorbose, and the like; disaccharides, such as lactose, sucrose, trehalose, cellobiose, and the like; polysaccharides, such as raffinose, melezitose, maltodextrins, dextrans, starches, and the like; and alditols, such as mannitol, xylitol, maltitol, lactitol, xylitol, sorbitol (glucitol), pyranosyl sorbitol, myoinositol, and
  • the excipient can also include an inorganic salt or buffer such as citric acid, sodium chloride, potassium chloride, sodium sulfate, potassium nitrate, sodium phosphate monobasic, sodium phosphate dibasic, and combinations thereof.
  • an inorganic salt or buffer such as citric acid, sodium chloride, potassium chloride, sodium sulfate, potassium nitrate, sodium phosphate monobasic, sodium phosphate dibasic, and combinations thereof.
  • the preparation may also include an antimicrobial agent for preventing or deterring microbial growth.
  • antimicrobial agents suitable for the present invention include benzalkonium chloride, benzethonium chloride, benzyl alcohol, cetylpyridinium chloride, chlorobutanol, phenol, phenylethyl alcohol, phenylmercuric nitrate, thimersol, and combinations thereof.
  • An antioxidant can be present in the preparation as well. Antioxidants are used to prevent oxidation, thereby preventing the deterioration of the conjugate or other components of the preparation. Suitable antioxidants for use in the present invention include, for example, ascorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, hypophosphorous acid, monothioglycerol, propyl gallate, sodium bisulfite, sodium formaldehyde sulfoxylate, sodium metabisulfite, and combinations thereof.
  • a surfactant may be present as an excipient.
  • exemplary surfactants include: polysorbates, such as “Tween 20” and “Tween 80,” and pluronics such as F68 and F88 (both of which are available from BASF, Mount Olive, New Jersey); sorbitan esters; lipids, such as phospholipids such as lecithin and other phosphatidylcholines, phosphatidylethanolamines (although preferably not in liposomal form), fatty acids and fatty esters; steroids, such as cholesterol; and chelating agents, such as EDTA, zinc and other such suitable cations.
  • acids or bases may be present as an excipient in the preparation.
  • acids that can be used include those acids selected from the group consisting of hydrochloric acid, acetic acid, phosphoric acid, citric acid, malic acid, lactic acid, formic acid, trichloroacetic acid, nitric acid, perchloric acid, phosphoric acid, sulfuric acid, fumaric acid, and combinations thereof.
  • Suitable bases include, without limitation, bases selected from the group consisting of sodium hydroxide, sodium acetate, ammonium hydroxide, potassium hydroxide, ammonium acetate, potassium acetate, sodium phosphate, potassium phosphate, sodium citrate, sodium formate, sodium sulfate, potassium sulfate, potassium fumerate, and combinations thereof.
  • the amount of the conjugate in the composition will vary depending on a number of factors, but will optimally be a therapeutically effective dose when the composition is stored in a unit dose container.
  • a therapeutically effective dose can be determined experimentally by repeated administration of increasing amounts of the conjugate in order to determine which amount produces a clinically desired endpoint.
  • the amount of any individual excipient in the composition will vary depending on the activity of the excipient and particular needs of the composition.
  • the optimal amount of any individual excipient is determined through routine experimentation, i.e., by preparing compositions containing varying amounts of the excipient (ranging from low to high), examining the stability and other parameters, and then determining the range at which optimal performance is attained with no significant adverse effects.
  • the excipient will be present in the composition in an amount of about 1% to about 99% by weight, preferably from about 5%-98% by weight, more preferably from about 15-95% by weight of the excipient, with concentrations less than 30% by weight most preferred.
  • compositions can take any number of forms and the invention is not limited in this regard.
  • exemplary preparations are most preferably in a form suitable for oral administration such as a tablet, caplet, capsule, gel cap, troche, dispersion, suspension, solution, elixir, syrup, lozenge, transdermal patch, spray, suppository, and powder.
  • Oral dosage forms are preferred for those conjugates that are orally active, and include tablets, caplets, capsules, gel caps, suspensions, solutions, elixirs, and syrups, and can also comprise a plurality of granules, beads, powders or pellets that are optionally encapsulated.
  • Such dosage forms are prepared using conventional methods known to those in the field of pharmaceutical formulation and described in the pertinent texts.
  • Tablets and caplets can be manufactured using standard tablet processing procedures and equipment. Direct compression and granulation techniques are preferred when preparing tablets or caplets containing the conjugates described herein.
  • the tablets and caplets will generally contain inactive, pharmaceutically acceptable carrier materials such as binders, lubricants, disintegrants, fillers, stabilizers, surfactants, coloring agents, and the like. Binders are used to impart cohesive qualities to a tablet, and thus ensure that the tablet remains intact.
  • Suitable binder materials include, but are not limited to, starch (including corn starch and pregelatinized starch), gelatin, sugars (including sucrose, glucose, dextrose and lactose), polyethylene glycol, waxes, and natural and synthetic gums, e.g., acacia sodium alginate, polyvinylpyrrolidone, cellulosic polymers (including hydroxypropyl cellulose, hydroxypropyl methylcellulose, methyl cellulose, microcrystalline cellulose, ethyl cellulose, hydroxyethyl cellulose, and the like), and Veegum.
  • Lubricants are used to facilitate tablet manufacture, promoting powder flow and preventing particle capping (i.e., particle breakage) when pressure is relieved.
  • Useful lubricants are magnesium stearate, calcium stearate, and stearic acid.
  • Disintegrants are used to facilitate disintegration of the tablet, and are generally starches, clays, celluloses, algins, gums, or crosslinked polymers.
  • Fillers include, for example, materials such as silicon dioxide, titanium dioxide, alumina, talc, kaolin, powdered cellulose, and microcrystalline cellulose, as well as soluble materials such as mannitol, urea, sucrose, lactose, dextrose, sodium chloride, and sorbitol.
  • Stabilizers as well known in the art, are used to inhibit or retard drug decomposition reactions that include, by way of example, oxidative reactions.
  • Capsules are also preferred oral dosage forms, in which case the conjugate-containing composition can be encapsulated in the form of a liquid or gel (e.g., in the case of a gel cap) or solid (including particulates such as granules, beads, powders or pellets).
  • Suitable capsules include hard and soft capsules, and are generally made of gelatin, starch, or a cellulosic material. Two-piece hard gelatin capsules are preferably sealed, such as with gelatin bands or the like.
  • parenteral formulations in the substantially dry form typically as a lyophilizate or precipitate, which can be in the form of a powder or cake
  • formulations prepared for injection which are typically liquid and requires the step of reconstituting the dry form of parenteral formulation.
  • suitable diluents for reconstituting solid compositions prior to injection include bacteriostatic water for injection, dextrose 5% in water, phosphate-buffered saline, Ringer's solution, saline, sterile water, deionized water, and combinations thereof.
  • compositions intended for parenteral administration can take the form of nonaqueous solutions, suspensions, or emulsions, each typically being sterile.
  • nonaqueous solvents or vehicles are propylene glycol, polyethylene glycol, vegetable oils, such as olive oil and corn oil, gelatin, and injectable organic esters such as ethyl oleate.
  • parenteral formulations described herein can also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents.
  • adjuvants such as preserving, wetting, emulsifying, and dispersing agents.
  • the formulations are rendered sterile by incorporation of a sterilizing agent, filtration through a bacteria-retaining filter, irradiation, or heat.
  • the conjugate can also be administered through the skin using conventional transdermal patch or other transdermal delivery system, wherein the conjugate is contained within a laminated structure that serves as a drug delivery device to be affixed to the skin.
  • the conjugate is contained in a layer, or "reservoir,” underlying an upper backing layer.
  • the laminated structure can contain a single reservoir, or it can contain multiple reservoirs.
  • the conjugate can also be formulated into a suppository for rectal administration.
  • a suppository base material which is (e.g., an excipient that remains solid at room temperature but softens, melts or dissolves at body temperature) such as coca butter (theobroma oil), polyethylene glycols, glycerinated gelatin, fatty acids, and combinations thereof.
  • Suppositories can be prepared by, for example, performing the following steps (not necessarily in the order presented): melting the suppository base material to form a melt; incorporating the conjugate (either before or after melting of the suppository base material); pouring the melt into a mold; cooling the melt (e.g., placing the melt-containing mold in a room temperature environment) to thereby form suppositories; and removing the suppositories from the mold.
  • the invention also provides a method for administering a conjugate as provided herein to a patient suffering from a condition that is responsive to treatment with the conjugate.
  • the method comprises administering, generally orally, a therapeutically effective amount of the conjugate (preferably provided as part of a pharmaceutical preparation).
  • Other modes of administration are also contemplated, such as pulmonary, nasal, buccal, rectal, sublingual, transdermal, and parenteral.
  • parenteral includes subcutaneous, intravenous, intra-arterial, intraperitoneal, intracardiac, intrathecal, and intramuscular injection, as well as infusion injections.
  • oligomers In instances where parenteral administration is utilized, it may be necessary to employ somewhat bigger oligomers than those described previously, with molecular weights ranging from about 500 to 30K Daltons (e.g., having molecular weights of about 500, 1000, 2000, 2500, 3000, 5000, 7500, 10000, 15000, 20000, 25000, 30000 or even more).
  • the method of administering may be used to treat any condition that can be remedied or prevented by administration of the particular conjugate.
  • Those of ordinary skill in the art appreciate which conditions a specific conjugate can effectively treat.
  • the actual dose to be administered will vary depend upon the age, weight, and general condition of the subject as well as the severity of the condition being treated, the judgment of the health care professional, and conjugate being administered.
  • Therapeutically effective amounts are known to those skilled in the art and/or are described in the pertinent reference texts and literature. Generally, a therapeutically effective amount will range from about 0.001 mg to 1000 mg, preferably in doses from 0.01 mg/day to 750 mg/day, and more preferably in doses from 0.10 mg/day to 500 mg/day.
  • the unit dosage of any given conjugate (again, preferably provided as part of a pharmaceutical preparation) can be administered in a variety of dosing schedules depending on the judgment of the clinician, needs of the patient, and so forth.
  • the specific dosing schedule will be known by those of ordinary skill in the art or can be determined experimentally using routine methods.
  • Exemplary dosing schedules include, without limitation, administration five times a day, four times a day, three times a day, twice daily, once daily, three times weekly, twice weekly, once weekly, twice monthly, once monthly, and any combination thereof. Once the clinical endpoint has been achieved, dosing of the composition is halted.
  • One advantage of administering the conjugates of the present invention is that a reduction in first pass metabolism may be achieved relative to the parent drug. Such a result is advantageous for many orally administered drugs that are substantially metabolized by passage through the gut. In this way, clearance of the conjugate can be modulated by selecting the oligomer molecular size, linkage, and position of covalent attachment providing the desired clearance properties.
  • One of ordinary skill in the art can determine the ideal molecular size of the oligomer based upon the teachings herein.
  • Preferred reductions in first pass metabolism for a conjugate as compared to the corresponding nonconjugated small drug molecule include : at least about 10%, at least about 20%, at least about 30; at least about 40; at least about 50%; at least about 60%, at least about 70%, at least about 80% and at least about 90%.
  • the invention provides a method for reducing the metabolism of an active agent.
  • the method comprises the steps of: providing monodisperse or bimodal conjugates, each conjugate comprised of a moiety derived from a small molecule drug covalently attached by a stable linkage to a water-soluble oligomer, wherein said conjugate exhibits a reduced rate of metabolism as compared to the rate of metabolism of the small molecule drug not attached to the water-soluble oligomer; and administering the conjugate to a patient.
  • administration is carried out via one type of administration selected from the group consisting of oral administration, transdermal administration, buccal administration, transmucosal administration, vaginal administration, rectal administration, parenteral administration, and pulmonary administration.
  • the conjugates are particularly useful when the small molecule drug is metabolized by a hepatic enzyme (e.g., one or more of the cytochrome P450 isoforms) and/or by one or more intestinal enzymes.
  • a hepatic enzyme e.g., one or more of the cytochrome P450 isoforms
  • intestinal enzymes e.g., one or more of the cytochrome P450 isoforms
  • PEG-Nalbuphine was prepared using a first approach. Schematically, the approach followed for this example is shown below.
  • Nalbuphine hydrochloride dihydrate 600 mg, from Sigma was dissolved in water (100 mL). Saturated aqueous K 2 CO 3 was added and then adjusted the pH to 9.3 with 1 N HCl solution, saturated with sodium chloride. The solution was extracted with dichloromethane (5 x 25 mL). The combined organic solution was washed with brine (100 mL), dried over Na 2 SO 4 , concentrated to dryness and dried under high vacuum to yield nalbuphine (483.4 mg, 97% recovery). The product was confirmed by 1 H-NMR in CDCl 3 .
  • Nalbuphine (28.5 mg, 0.08 mmol) was dissolved in a mixture of acetone (2 mL) and toluene (1.5 mL). Potassium carbonate (21 mg, 0.15 mmol) was added, followed by an addition of mPEG 3 -Br (44.5 mg, 0.20 mmol) at room temperature. The resulting mixture was stirred at room temperature for 27.5 hours. More potassium carbonate (24 mg, 0.17 mmol) was added. The mixture was heated with CEM microwave such that 60 °C for 20 minutes was achieved, and then such that 100 °C for 30 minutes was achieved. DMF (0.2 mL) was added. The mixture was heated with microwave at 60 °C for 20 minutes, at 100 °C for 30 minutes.
  • the reaction was concentrated to remove the organic solvents, the residue was mixed with water (10 mL), extracted with dichloromethane (4 x 15 mL). The combined organic solution was washed with brine, dried over Na 2 SO 4 , concentrated.
  • the crude product was checked with HPLC and LC-MS. The residue was mixed again with water (10 mL), adjusted the pH to 2.3 with 1N HCl, washed with dichloromethane (2 x 15 mL). The aqueous solution was adjusted to pH 10.4 with 0.2 N NaOH, extracted with dichloromethane (4 x 15 mL). The combined organic solution was washed with brine, dried over Na 2 SO 4 , concentrated.
  • a mixture of nalbuphine (60 mg, 0.17 mmol) and mPEG 8 -Br (105.7 mg, 0.24 mmol) in the presence of potassium carbonate (40.8 mg, 0.30 mmol) in toluene/DMF (3 mL/0.3 mL) was heated with CEM microwave such that 100 °C for 30 minutes was achieved. Then acetone (1 mL) was added. After the mixture was heated with CEM microwave such that 100 °C for 90 minutes was achieved, more of K 2 CO 3 (31 mg, 0.22 mmol) and mPEG 8 -Br (100 mg, 0.22 mmol) were added. The mixture was heated with CEM microwave such that 100 °C for 60 minutes was achieved.
  • PEG-Nalbuphine was prepared using a second approach. Schematically, the approach followed for this example is shown below.
  • a 20-mL vial was placed with 3-O-MEM-nalbuphine (3) (85 mg, 0.19 mmol) and toluene (15 mL). The mixture was concentrated to remove 7 mL of toluene. Anhydrous DMF (0.2 mL) was added. The vial was flashed with nitrogen. NaH (60% dispersion in mineral oil, 21 mg, 0.53 mmol) was added, followed by an addition of mPEG 3 -OMs (94 mg, 0.39 mmol). After the resulting mixture was heated at 45 °C for 22.5 hours, more of NaH (22 mg, 0.55 mmol) was added.
  • 6-O-mPEG 4 -3-O-MEM-nalbuphine (4) (214.4 mg) was stirred in 2 M HCl in methanol (30 mL) at room temperature for 6 hours. The mixture was diluted with water (5 mL), and concentrated to removed the methanol. The aqueous solution was adjusted to 9.17 with 1 N NaOH, extracted with dichloromethane (4 x 25 mL). The combined organic solution was washed with brine, dried over Na 2 SO 4 , and concentrated.
  • 3-O-MEM-nalbuphine (3) (77.6 mg, 0.17 mmol) and mPEG 6 -OMs (199 mg, 0.53 mmol) was dissolved in toluene (20 mL). The mixture was concentrated to remove about 12 mL of toluene. Anhydrous DMF (0.2 mL) was added, followed by an addition of NaH (60% dispersion in mineral oil, 41 mg, 1.03 mmol). After the resulting mixture was heated at 45 °C for 23 hours, more of NaH (46 mg) was added. The mixture was heated at 45 °C for another 24 hours. When the mixture was cooled to room temperature, saturated NaCl aqueous solution (5 mL) was added to quench the reaction. The mixture was diluted with water (10 mL), extracted with EtOAc (4 x 15 mL). The combined organic solution was washed with brine, dried over Na 2 SO 4 , concentrated. The residue was directly used for the next step.
  • PEG-Nalbuphine was prepared using a third approach. Schematically, the approach followed for this example is shown below.
  • TrO-PEG 5 -OH Following a similar procedure for the preparation of TrO-PEG 5 -OH, other TrO-PEG n -OH were synthesized from the corresponding PEG n -di-OH.
  • TrO-PEG 5 -OMs Following a similar procedure for the preparation of TrO-PEG 5 -OMs, other TrO-PEG n -OMs were synthesized from the corresponding TrO-PEG n -OH.
  • PEG-U50488 can be prepared following the approach schematically shown below. Conventional organic synthetic techniques are used in carrying out the approach.
  • PEG-U69593 can be prepared following the approach schematically shown below. Conventional organic synthetic techniques are used in carrying out the approach.
  • Conjugates of opioid agonists other than nalbuphine, U50488 and U69593 can be prepared wherein the general synthetic scheme and procedures set forth in Example 1 can be followed except that an opioid agonist of Formula I is substituted for nalbuphine, U50488 and U69593.
EP13155667.2A 2007-03-12 2008-03-12 Conjugués d'agoniste opioïde-oligomère Not-in-force EP2620163B1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP17151793.1A EP3222293B1 (fr) 2007-03-12 2008-03-12 Conjugués d'agoniste opioïde-oligomère

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US90638707P 2007-03-12 2007-03-12
EP08742082.4A EP2134371B1 (fr) 2007-03-12 2008-03-12 Conjugués d'oligomère-agoniste opioïde

Related Parent Applications (3)

Application Number Title Priority Date Filing Date
EP08742082.4A Division-Into EP2134371B1 (fr) 2007-03-12 2008-03-12 Conjugués d'oligomère-agoniste opioïde
EP08742082.4A Division EP2134371B1 (fr) 2007-03-12 2008-03-12 Conjugués d'oligomère-agoniste opioïde
EP08742082.4 Division 2008-03-12

Related Child Applications (2)

Application Number Title Priority Date Filing Date
EP17151793.1A Division EP3222293B1 (fr) 2007-03-12 2008-03-12 Conjugués d'agoniste opioïde-oligomère
EP17151793.1A Division-Into EP3222293B1 (fr) 2007-03-12 2008-03-12 Conjugués d'agoniste opioïde-oligomère

Publications (2)

Publication Number Publication Date
EP2620163A1 true EP2620163A1 (fr) 2013-07-31
EP2620163B1 EP2620163B1 (fr) 2017-05-03

Family

ID=39590911

Family Applications (4)

Application Number Title Priority Date Filing Date
EP08742082.4A Active EP2134371B1 (fr) 2007-03-12 2008-03-12 Conjugués d'oligomère-agoniste opioïde
EP17151793.1A Active EP3222293B1 (fr) 2007-03-12 2008-03-12 Conjugués d'agoniste opioïde-oligomère
EP13155670.6A Withdrawn EP2623123A1 (fr) 2007-03-12 2008-03-12 Conjugués d'agoniste opioïde avec un oligomère
EP13155667.2A Not-in-force EP2620163B1 (fr) 2007-03-12 2008-03-12 Conjugués d'agoniste opioïde-oligomère

Family Applications Before (3)

Application Number Title Priority Date Filing Date
EP08742082.4A Active EP2134371B1 (fr) 2007-03-12 2008-03-12 Conjugués d'oligomère-agoniste opioïde
EP17151793.1A Active EP3222293B1 (fr) 2007-03-12 2008-03-12 Conjugués d'agoniste opioïde-oligomère
EP13155670.6A Withdrawn EP2623123A1 (fr) 2007-03-12 2008-03-12 Conjugués d'agoniste opioïde avec un oligomère

Country Status (18)

Country Link
EP (4) EP2134371B1 (fr)
JP (2) JP5420427B2 (fr)
KR (1) KR101568428B1 (fr)
CN (1) CN101646464B (fr)
AU (1) AU2008226822B2 (fr)
CA (1) CA2679479C (fr)
CY (1) CY1117368T1 (fr)
DK (1) DK2134371T3 (fr)
ES (1) ES2534741T3 (fr)
HR (1) HRP20150315T1 (fr)
IL (1) IL200845A (fr)
ME (1) ME02176B (fr)
MX (1) MX2009009851A (fr)
PL (1) PL2134371T3 (fr)
PT (1) PT2134371E (fr)
RS (1) RS53937B1 (fr)
SI (1) SI2134371T1 (fr)
WO (1) WO2008112288A2 (fr)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10512644B2 (en) 2007-03-12 2019-12-24 Inheris Pharmaceuticals, Inc. Oligomer-opioid agonist conjugates
US8173666B2 (en) 2007-03-12 2012-05-08 Nektar Therapeutics Oligomer-opioid agonist conjugates
KR101660996B1 (ko) * 2008-09-16 2016-09-28 넥타르 테라퓨틱스 남용에 대한 낮은 잠재성을 갖는 페길화 오피오이드
CN105503889B (zh) * 2009-07-21 2019-12-24 尼克塔治疗公司 低聚物-阿片样激动剂轭合物
MX2012000980A (es) * 2009-07-21 2012-06-12 Nektar Therapeutics Conjugados de agonistas de oligómeros-opioides.
US20130023553A1 (en) * 2010-01-12 2013-01-24 Nektar Therapeutics Pegylated opioids with low potential for abuse and side effects
US20130022669A1 (en) * 2010-03-30 2013-01-24 Spago Imaging Ab Branched, compact polyethyleneglycol derivatives
CA2819280C (fr) 2010-11-30 2016-10-04 Research In Motion Limited Reselection de cellule dans un reseau de telecommunication cellulaire
EP2776046A1 (fr) 2011-11-07 2014-09-17 Nektar Therapeutics Compositions, formes galéniques et coadministration d'un composé agoniste opioïde et d'un composé analgésique
US10525054B2 (en) 2011-11-07 2020-01-07 Inheris Biopharma, Inc. Compositions, dosage forms, and co-administration of an opioid agonist compound and an analgesic compound
ME02971B (fr) * 2012-10-30 2018-07-20 Sels solides de alpha-6-mpeg6-o-hydroxycodone comme opioid agonistes et leurs utilisation
JP6577943B2 (ja) * 2013-06-28 2019-09-18 ネクター セラピューティクス κオピオイド作動薬及びその使用
CN107019803B (zh) * 2016-01-29 2020-09-15 北京键凯科技股份有限公司 具有低成瘾作用的聚乙二醇化阿片样物质
CN107033154B (zh) 2016-02-02 2020-02-04 上海瀚迈生物医药科技有限公司 阿片受体拮抗剂缀合物及其应用
CN108210933B (zh) * 2016-12-22 2021-05-04 北京键凯科技股份有限公司 一种地佐辛与聚乙二醇的结合物
US11091497B2 (en) * 2017-12-08 2021-08-17 The Rockefeller University Pyrano[3,4-b]pyrazine kappa opioid receptor ligands for treating addiction, pruritus, pain, and inflammation
CA3095299A1 (fr) 2018-03-29 2019-10-03 Nof Corporation Procede de purification de polyethylene glycol monodisperse contenant un groupe trityle

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003032990A2 (fr) * 2001-10-18 2003-04-24 Nektar Therapeutics Al, Corporation Conjugues polymeres d'antagonistes opioides
WO2004039317A2 (fr) * 2002-10-25 2004-05-13 Euro-Celtique S.A. Analogues et promedicaments de buprenorphine
WO2004082620A2 (fr) * 2003-03-13 2004-09-30 Controlled Chemicals, Inc. Composes et methodes pour reduire l'abus potentiel d'un medicament et pour en prolonger la duree d'action
WO2005058367A2 (fr) * 2003-12-16 2005-06-30 Nektar Therapeutics Al, Corporation Petites molecules chimiquement modifiees
WO2008036980A1 (fr) * 2006-09-22 2008-03-27 Alltranz Inc. Promédicaments d'opioïdes administrables de façon transdermique, compositions empêchant l'usage abusif de ceux-ci et procédés d'utilisation de promédicaments d'opioïdes
WO2008057579A2 (fr) * 2006-11-07 2008-05-15 Nektar Therapeutics Al, Corporation Formes dosifiées et co-administration d'un agoniste opioïde et d'un antagoniste opioïde

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2654756A (en) 1949-10-20 1953-10-06 Mallinckrodt Chemical Works Process of preparing codeinone, dihydrocodeinone, and dihydromorphinone
US2628962A (en) 1949-10-20 1953-02-17 Mallinckrodt Chemical Works Method for preparing dihydrocodeinone, dihydromorphinone, and codeinone
US2649454A (en) 1951-08-20 1953-08-18 Univ California Method for preparing dihydromorphinone, dihydrocodeinone, and dihydropseudocodeinone
US2806033A (en) 1955-08-03 1957-09-10 Lewenstein Morphine derivative
US5475019A (en) * 1993-02-08 1995-12-12 East Carolina University Method of treating anxiety-related disorders with 2-aminocycloaliphatic amide compounds
US5672662A (en) 1995-07-07 1997-09-30 Shearwater Polymers, Inc. Poly(ethylene glycol) and related polymers monosubstituted with propionic or butanoic acids and functional derivatives thereof for biotechnical applications
US6835802B2 (en) 2001-06-04 2004-12-28 Nobex Corporation Methods of synthesizing substantially monodispersed mixtures of polymers having polyethylene glycol moieties
WO2003058367A1 (fr) 2002-01-11 2003-07-17 Valquest Limited Navette de melangeur de flux
EP1487813B1 (fr) * 2002-02-22 2006-08-30 Bayer Pharmaceuticals Corporation Derives de benzofurane et de benzothiophene utilises dans le traitement de troubles hyperproliferatifs

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003032990A2 (fr) * 2001-10-18 2003-04-24 Nektar Therapeutics Al, Corporation Conjugues polymeres d'antagonistes opioides
WO2004039317A2 (fr) * 2002-10-25 2004-05-13 Euro-Celtique S.A. Analogues et promedicaments de buprenorphine
WO2004082620A2 (fr) * 2003-03-13 2004-09-30 Controlled Chemicals, Inc. Composes et methodes pour reduire l'abus potentiel d'un medicament et pour en prolonger la duree d'action
WO2005058367A2 (fr) * 2003-12-16 2005-06-30 Nektar Therapeutics Al, Corporation Petites molecules chimiquement modifiees
WO2008036980A1 (fr) * 2006-09-22 2008-03-27 Alltranz Inc. Promédicaments d'opioïdes administrables de façon transdermique, compositions empêchant l'usage abusif de ceux-ci et procédés d'utilisation de promédicaments d'opioïdes
WO2008057579A2 (fr) * 2006-11-07 2008-05-15 Nektar Therapeutics Al, Corporation Formes dosifiées et co-administration d'un agoniste opioïde et d'un antagoniste opioïde

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
EREZ M ET AL: "Narcotic antagonistic potency of bivalent ligands which contain beta-naltrexamine. Evidence for bridging between proximal recognition sites", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. WASHINGTON, vol. 25, no. 7, 1 July 1982 (1982-07-01), pages 847 - 849, XP002334914, ISSN: 0022-2623 *
FUKUDA KAZUHIKO ET AL: "Partial agonistic activity of naloxone on the opioid receptors expressed from complementary deoxyribonucleic acids in Chinese hamster ovary cells", ANESTHESIA AND ANALGESIA, vol. 87, no. 2, August 1998 (1998-08-01), pages 450 - 455, XP002488162, ISSN: 0003-2999 *
OKUDA T ET AL: "PROLONGED ANTINOCICEPTIVE EFFECT AFTER EPIDURAL INJECTION OF POLYETHYLENEGLYCOL-MORPHINE COMPOSITES IN RATS", MASUI - JAPANESE JOURNAL OF ANESTHESIOLOGY, TOKYO, JP, vol. 45, no. 5, 1 January 1996 (1996-01-01), pages 571 - 575, XP008035141, ISSN: 0021-4892 *

Also Published As

Publication number Publication date
PT2134371E (pt) 2015-04-16
JP2010521466A (ja) 2010-06-24
CY1117368T1 (el) 2017-04-26
RS53937B1 (en) 2015-08-31
CN101646464B (zh) 2013-05-29
HRP20150315T1 (hr) 2015-04-24
DK2134371T3 (en) 2015-04-27
IL200845A (en) 2015-08-31
EP2623123A1 (fr) 2013-08-07
AU2008226822A1 (en) 2008-09-18
ES2534741T3 (es) 2015-04-28
KR101568428B1 (ko) 2015-11-11
CN101646464A (zh) 2010-02-10
PL2134371T3 (pl) 2015-06-30
JP5420427B2 (ja) 2014-02-19
CA2679479A1 (fr) 2008-09-18
CA2679479C (fr) 2015-10-06
EP2134371B1 (fr) 2015-01-14
KR20090118952A (ko) 2009-11-18
JP5877403B2 (ja) 2016-03-08
IL200845A0 (en) 2010-05-17
EP3222293A1 (fr) 2017-09-27
WO2008112288A2 (fr) 2008-09-18
SI2134371T1 (sl) 2015-04-30
EP2620163B1 (fr) 2017-05-03
EP3222293B1 (fr) 2019-11-27
AU2008226822B2 (en) 2013-08-01
EP2134371A2 (fr) 2009-12-23
MX2009009851A (es) 2009-09-24
ME02176B (me) 2015-10-20
JP2013151563A (ja) 2013-08-08
WO2008112288A3 (fr) 2008-12-24

Similar Documents

Publication Publication Date Title
CA2679479C (fr) Conjugues d'oligomere-agoniste opioide
US10143690B2 (en) Oligomer-opioid agonist conjugates
US20160243111A1 (en) Pegylated Opioids with Low Potential for Abuse
EP2628489B1 (fr) Conjugués de PEG oligomère-fentanyl
US20200078353A1 (en) Oligomer-opioid agonist conjugates

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

AC Divisional application: reference to earlier application

Ref document number: 2134371

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA MK RS

17P Request for examination filed

Effective date: 20140130

RBV Designated contracting states (corrected)

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

17Q First examination report despatched

Effective date: 20151123

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTG Intention to grant announced

Effective date: 20160922

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

RIN1 Information on inventor provided before grant (corrected)

Inventor name: RIGGS-SAUTHIER, JENNIFER

Inventor name: DENG, BO-LIANG

Inventor name: RILEY, TIMOTHY, ANDREW

REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 602008050151

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: A61K0047480000

Ipc: A61K0047500000

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AC Divisional application: reference to earlier application

Ref document number: 2134371

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 47/50 20170101AFI20170328BHEP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 889237

Country of ref document: AT

Kind code of ref document: T

Effective date: 20170515

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602008050151

Country of ref document: DE

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20170503

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 889237

Country of ref document: AT

Kind code of ref document: T

Effective date: 20170503

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170804

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170803

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170903

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170803

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 11

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602008050151

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20180206

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20180331

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180312

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180312

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180331

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180331

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180331

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180312

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20200130

Year of fee payment: 13

Ref country code: DE

Payment date: 20200225

Year of fee payment: 13

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20080312

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20170503

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20200124

Year of fee payment: 13

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602008050151

Country of ref document: DE

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20210312

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20211001

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210331

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20210312